1
|
Boucher DM, Robichaud S, Lorant V, Leon JS, Suliman I, Rasheed A, Susser LI, Emerton C, Geoffrion M, De Jong E, Bowdish DM, Aikawa M, Aikawa E, Singh SA, Rayner KJ, Ouimet M. Age-Related Impairments in Immune Cell Efferocytosis and Autophagy Hinder Atherosclerosis Regression. Arterioscler Thromb Vasc Biol 2025; 45:481-495. [PMID: 39945065 PMCID: PMC11936474 DOI: 10.1161/atvbaha.124.321662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 01/23/2025] [Indexed: 02/28/2025]
Abstract
BACKGROUND Aging is a well-established risk factor for the development and progression of atherosclerosis, but the molecular mechanisms underlying this relationship remain poorly defined, and its role in atherosclerosis regression is unknown. To uncover age-related alterations that may impair atherosclerosis regression, we investigated the response of young and old macrophages to atherogenic lipoproteins in vitro and in vivo. METHODS Metabolic and proteomic studies were performed in vitro using macrophages differentiated from the bone marrow of young or old mice. To test the role of immune cell aging in atherosclerosis regression, bone marrow from young and old donors was transplanted into irradiated young recipient mice expressing gain-of-function AAV-PCSK9 (adeno-associated virus-proprotein convertase subtilisin/kexin type 9). Following 14 weeks of Western diet feeding, atherosclerosis regression was induced by switching to a standard laboratory diet for 4 weeks. RESULTS Compared with young macrophages, old macrophages accumulated more lipid droplets upon lipid loading with the pro-atherogenic lipoprotein aggregated LDL (low-density lipoprotein), accompanied by a failure to proportionally induce autophagy and cholesterol efflux. Proteomic analysis of bone marrow-derived macrophages revealed that pathways related to endocytosis, engulfment, and phagocytosis were downregulated in old lipid-loaded macrophages. Functional studies confirmed a reduction in efferocytic capacity in old macrophages. In recipient mice transplanted with old bone marrow, atherosclerosis regression was impaired, as evidenced by inefficient resolution of circulating inflammatory cell levels, reduced activation of plaque autophagy and apoptotic cell clearance, and persistent plaque CD45+ and CD68+ content. CONCLUSIONS Aging impairs macrophage function through reduced efferocytosis and autophagy activation, limiting atherosclerosis regression. These results highlight the need to better define the mechanisms linking aging to atherosclerosis to develop targeted therapies for the aging population.
Collapse
Affiliation(s)
- Dominique M. Boucher
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Sabrina Robichaud
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Victoria Lorant
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Jonathan S. Leon
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Issraa Suliman
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Adil Rasheed
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Leah I. Susser
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Christina Emerton
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Michele Geoffrion
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Erica De Jong
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada (E.D.J., D.M.E.B.)
- Firestone Institute for Respiratory Health, St. Joseph’s Healthcare Hamilton, ON, Canada (E.D.J., D.M.E.B.)
| | - Dawn M.E. Bowdish
- McMaster Immunology Research Centre, McMaster University, Hamilton, ON, Canada (E.D.J., D.M.E.B.)
- Firestone Institute for Respiratory Health, St. Joseph’s Healthcare Hamilton, ON, Canada (E.D.J., D.M.E.B.)
| | - Masanori Aikawa
- Cardiovascular Division, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences (M.A., E.A., S.A.S.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Cardiovascular Division, Department of Medicine, Center for Excellence in Vascular Biology (M.A., E.A., S.A.S.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Elena Aikawa
- Cardiovascular Division, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences (M.A., E.A., S.A.S.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Cardiovascular Division, Department of Medicine, Center for Excellence in Vascular Biology (M.A., E.A., S.A.S.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Sasha A. Singh
- Cardiovascular Division, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences (M.A., E.A., S.A.S.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Cardiovascular Division, Department of Medicine, Center for Excellence in Vascular Biology (M.A., E.A., S.A.S.), Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Katey J. Rayner
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| | - Mireille Ouimet
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, ON, Canada (D.M.B., S.R., V.L., I.S., A.R., L.I.S., K.J.R., M.O.)
- University of Ottawa Heart Institute, ON, Canada (D.M.B., S.R., V.L., J.S.L., I.S., A.R., L.I.S., C.E., M.G., K.J.R., M.O.)
| |
Collapse
|
2
|
Su Y, Liu L, Lin C, Deng D, Li Y, Huang M, Wang Y, Ling K, Wang H, Chen Q, Huang G. Enhancing cancer therapy: advanced nanovehicle delivery systems for oridonin. Front Pharmacol 2024; 15:1476739. [PMID: 39691396 PMCID: PMC11649421 DOI: 10.3389/fphar.2024.1476739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024] Open
Abstract
Oridonin (ORI), an ent-kaurane diterpenoid derived from Rabdosia rubescens (Hemsl.) H.Hara, serves as the primary bioactive component of this plant. It demonstrates a broad spectrum of therapeutic activities, including moderate to potent anticancer properties, alongside anti-inflammatory, antibacterial, antifibrotic, immunomodulatory, and neuromodulatory effects, thus influencing diverse biological processes. However, its clinical potential is significantly constrained by poor aqueous solubility and limited bioavailability. In alignment with the approach of developing drug candidates from natural compounds, various strategies, such as structural modification and nanocarrier systems, have been employed to address these challenges. This review provides an overview of ORI-based nano-delivery systems, emphasizing their potential to improve the clinical applicability of oridonin in oncology. Although some progress has been made in advancing ORI nano-delivery research, it remains insufficient for clinical implementation, necessitating further investigation.
Collapse
Affiliation(s)
- Yilin Su
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Lisha Liu
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Chongyang Lin
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Dashi Deng
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Yunfei Li
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Mou Huang
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Yu Wang
- Institute of Pain, The Affiliated Hospital of Southwest Jiaotong University, The Chengdu Third People’s Hospital, Chengdu, China
| | - Kangqiu Ling
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Haobing Wang
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Qiyu Chen
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Guixiao Huang
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| |
Collapse
|
3
|
Liu P, Wu X, Lv H, Huang J, Gu T, Liu D, Xu Y. Oridonin alleviates cigarette smoke-induced nasal polyp formation by promoting autophagy. Biomed Pharmacother 2024; 180:117547. [PMID: 39405900 DOI: 10.1016/j.biopha.2024.117547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 11/14/2024] Open
Abstract
Previous studies have indicated that oridonin is a promising candidate for therapeutic intervention in a range of inflammatory diseases. The objective of this study was to investigate the protective mechanism of oridonin in chronic rhinosinusitis with nasal polyp (CRSwNP). In nasal polyp (NP) mice model, cigarette smoke (CS) induced polypoid changes compared to previous modeling methods. Compared with CS-treated mice, oridonin reduced polypoid changes, goblet cell count, and promoted the expression of tight junction proteins (ZO-1, occludin, claudin-1) and production of autophagosomes. Following treatment with oridonin, the levels of OVA-specific IgE, IL-6, IFN-γ, IL-5, IL-13 and IL-17A in serum were observed to decrease; the levels of TGF-β1, matrix metalloproteinase 2 (MMP2), MMP7, MMP9 and MMP12 levels in nasal lavage fluid were reduced, while tissue inhibitor of metalloproteinase-1 (TIMP-1) levels were increased. Furthermore, the aforementioned alterations in the mouse model were reversed by 3-methyladenine (3-MA), an autophagy inhibitor. In vitro, cigarette smoke extract (CSE) was observed to decrease the expression of tight junction proteins, the production of autophagosomes, and to reduce the expression of LC3-II and Beclin-1, accompanied by an increase in P62 expression. In addition, oridonin was observed to reverse CSE-induced epithelial barrier damage, and was associated with autophagy and the PI3K/AKT/mTOR pathway. In conclusion, oridonin was demonstrated to improve the damage of the nasal epithelial barrier induced by CS through the promotion of autophagy, which may represent a novel therapeutic option for the treatment of CRSwNP.
Collapse
Affiliation(s)
- Peiqiang Liu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China; Department of Rhinology and Allergy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaomin Wu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China; Department of Rhinology and Allergy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hao Lv
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China; Department of Rhinology and Allergy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jingyu Huang
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China; Department of Rhinology and Allergy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tian Gu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China; Department of Rhinology and Allergy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Duo Liu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China; Department of Rhinology and Allergy, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yu Xu
- Department of Otolaryngology-Head and Neck Surgery, Renmin Hospital of Wuhan University, Wuhan, China; Department of Rhinology and Allergy, Renmin Hospital of Wuhan University, Wuhan, China; Hubei Province Key Laboratory of Allergy and Immunology, Wuhan, China.
| |
Collapse
|
4
|
Kim JH, Song JW, Kim YH, Kim HJ, Kim RH, Park YH, Nam HS, Kang DO, Yoo H, Park K, Kim JW. Multimodal Imaging-Assisted Intravascular Theranostic Photoactivation on Atherosclerotic Plaque. Circ Res 2024; 135:e114-e132. [PMID: 38989585 DOI: 10.1161/circresaha.123.323970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 07/12/2024]
Abstract
BACKGROUND Atherosclerosis is a chronic inflammatory disease causing a fatal plaque rupture, and its key aspect is a failure to resolve inflammation. We hypothesize that macrophage-targeted near-infrared fluorescence emitting photoactivation could simultaneously assess macrophage/lipid-rich plaques in vivo and facilitate inflammation resolution. METHODS We fabricated a Dectin-1-targeted photoactivatable theranostic agent through the chemical conjugation of the near-infrared fluorescence-emitting photosensitizer chlorin e6 and the Dectin-1 ligand laminarin (laminarin-chlorin e6 [LAM-Ce6]). Intravascular photoactivation by a customized fiber-based diffuser after administration of LAM-Ce6 effectively reduced inflammation in the targeted plaques of atherosclerotic rabbits in vivo as serially assessed by dual-modal optical coherence tomography-near-infrared fluorescence structural-molecular catheter imaging after 4 weeks. RESULTS The number of apoptotic macrophages peaked at 1 day after laser irradiation and then resolved until 4 weeks. Autophagy was strongly augmented 1 hour after the light therapy, with the formation of autophagolysosomes. LAM-Ce6 photoactivation increased the terminal deoxynucleotidyl transferase dUTP (deoxyuridine triphosphate) nick end labeling/RAM11 (rabbit monocyte/macrophage antibody)- and MerTK (c-Mer tyrosine kinase)-positive cells in the plaques, suggesting enhanced efferocytosis. In line with inflammation resolution, photoactivation reduced the plaque burden through fibrotic replacement via the TGF (transforming growth factor)-β/CTGF (connective tissue growth factor) pathway. CONCLUSIONS Optical coherence tomography-near-infrared fluorescence imaging-guided macrophage Dectin-1-targetable photoactivation could induce the transition of macrophage/lipid-rich plaques into collagen-rich lesions through autophagy-mediated inflammation resolution and TGF-β-dependent fibrotic replacement. This novel strategy offers a new opportunity for the catheter-based theranostic strategy.
Collapse
Affiliation(s)
- Jin Hyuk Kim
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea (J.H.K., J.W.K.)
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital (J.H.K., J.W.S., H.J.K., R.H.K., Y.H.P., D.O.K., J.W.K.)
| | - Joon Woo Song
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital (J.H.K., J.W.S., H.J.K., R.H.K., Y.H.P., D.O.K., J.W.K.)
| | - Yeon Hoon Kim
- Department of Mechanical Engineering, KAIST, Daejeon, Korea (Y.H.K., H.S.N., H.Y.)
| | - Hyun Jung Kim
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital (J.H.K., J.W.S., H.J.K., R.H.K., Y.H.P., D.O.K., J.W.K.)
| | - Ryeong Hyun Kim
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital (J.H.K., J.W.S., H.J.K., R.H.K., Y.H.P., D.O.K., J.W.K.)
| | - Ye Hee Park
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital (J.H.K., J.W.S., H.J.K., R.H.K., Y.H.P., D.O.K., J.W.K.)
| | - Hyeong Soo Nam
- Department of Mechanical Engineering, KAIST, Daejeon, Korea (Y.H.K., H.S.N., H.Y.)
| | - Dong Oh Kang
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital (J.H.K., J.W.S., H.J.K., R.H.K., Y.H.P., D.O.K., J.W.K.)
| | - Hongki Yoo
- Department of Mechanical Engineering, KAIST, Daejeon, Korea (Y.H.K., H.S.N., H.Y.)
| | - Kyeongsoon Park
- Department of Systems Biotechnology, Chung-Ang University, Anseong, Gyeonggi, Korea (K.P.)
| | - Jin Won Kim
- BK21 Graduate Program, Department of Biomedical Sciences, Korea University College of Medicine, Seoul, Korea (J.H.K., J.W.K.)
- Multimodal Imaging and Theranostic Laboratory, Cardiovascular Center, Korea University Guro Hospital (J.H.K., J.W.S., H.J.K., R.H.K., Y.H.P., D.O.K., J.W.K.)
| |
Collapse
|
5
|
Meiling L, Yiran C, Xiaoli S, Kaihui C, Toshihiko H, Kikuji I, Kazunori M, Hattori S, Fujisaki H, Liu W, Ikejima T. Gelatin but not type I collagen promotes bacteria phagocytosis in PMA-treated U937 human lymphoma cells. Connect Tissue Res 2024; 65:170-185. [PMID: 38526028 DOI: 10.1080/03008207.2024.2330693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 03/09/2024] [Indexed: 03/26/2024]
Abstract
PURPOSE Besides comprising scaffolding, extracellular matrix components modulate many biological processes including inflammation and cell differentiation. We previously found precoating cell plates with extracellular matrix collagen I, or its denatured product gelatin, causes aggregation of macrophage-like human lymphoma U937 cells, which are induced to differentiation by phorbol myristate treatment. In the present study, we investigated the influence of gelatin or collagen I precoating on the bacteria phagocytosis in PMA-stimulated U937 cells. MATERIALS AND METHODS Colony forming units of phagocytosed bacteria, Giemsa-staining of cells with phagocytosed bacteria, confocal microscopic and flow cytometric analysis of cells with phagocytosed FITC-labeled bacteria and non-bioactive latex beats were conducted. RESULTS Gelatin precoating enhances the phagocytosis of both Gram-negative and positive bacteria, as shown by the increased colony forming units of bacteria phagocytosed by cells, and increased intracellular bacteria observed after Giemsa-staining. But collagen I has no marked influence. Confocal microscopy reveals that both live and dead FITC-bacteria were phagocytosed more in the cells with gelatin-coating but not collagen-coating. Of note, both gelatin and collagen I coating had no influence on the phagocytosis of non-bioactive latex beads. Since gelatin-coating increases autophagy but collagen I has no such impact, we are curious about the role of autophagy. Inhibiting autophagy reduced the phagocytosis of bacteria, in cells with gelatin-coating, while stimulating autophagy enhanced phagocytosis. CONCLUSION This study finds the bacteria-phagocytosis stimulatory effect of gelatin in PMA-treated U937 cells and reveals the positive regulatory role of autophagy, predicting the potential use of gelatin products in anti-bacterial therapy.
Collapse
Affiliation(s)
- Li Meiling
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Chen Yiran
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Sun Xiaoli
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Chen Kaihui
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Hayashi Toshihiko
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
- Nippi Research Institute of Biomatrix, Nippi Inc., Toride, Ibaraki, Japan
| | - Itoh Kikuji
- Biochemical Center, Japan SLC Inc., Shizuoka, Japan
| | - Mizuno Kazunori
- Nippi Research Institute of Biomatrix, Nippi Inc., Toride, Ibaraki, Japan
| | - Shunji Hattori
- Nippi Research Institute of Biomatrix, Nippi Inc., Toride, Ibaraki, Japan
| | - Hitomi Fujisaki
- Nippi Research Institute of Biomatrix, Nippi Inc., Toride, Ibaraki, Japan
| | - Weiwei Liu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Takashi Ikejima
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
- Key Laboratory of Computational Chemistry-Based Natural Antitumor Drug Research & Development, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| |
Collapse
|
6
|
Bae SH, Kim JH, Park TH, Lee K, Lee BI, Jang H. BMS794833 inhibits macrophage efferocytosis by directly binding to MERTK and inhibiting its activity. Exp Mol Med 2022; 54:1450-1460. [PMID: 36056187 PMCID: PMC9534909 DOI: 10.1038/s12276-022-00840-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/13/2022] [Accepted: 06/24/2022] [Indexed: 06/15/2023] Open
Abstract
Myeloid epithelial reproductive proto-oncogene tyrosine kinase (MERTK) plays an essential role in modulating cancer immune tolerance by regulating macrophage efferocytosis. Studies are underway to develop small-molecule chemicals that inhibit MERTK as cancer immunotherapeutic agents, but these efforts are in their early stages. This study identified BMS794833, whose primary targets are MET and VEGFR2, as a potent MERTK inhibitor and developed a real-time efferocytosis monitoring system. The X-ray cocrystal structure revealed that BMS794833 was in contact with the ATP-binding pocket and the allosteric back pocket, rendering MERTK inactive. Homogeneous time-resolved fluorescence kinetic and Western blotting analyses showed that BMS794833 competitively inhibited MERTK activity in vitro and inhibited the autophosphorylation of MERTK in macrophages. We developed a system to monitor MERTK-dependent efferocytosis in real time, and using this system, we confirmed that BMS794833 significantly inhibited the efferocytosis of differentiated macrophages. Finally, BMS794833 significantly inhibited efferocytosis in vivo in a mouse model. These data show that BMS794833 is a type II MERTK inhibitor that regulates macrophage efferocytosis. In addition, the real-time efferocytosis monitoring technology developed in this study has great potential for future applications.
Collapse
Affiliation(s)
- Seung-Hyun Bae
- Reasearch Institute, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Jung-Hoon Kim
- Reasearch Institute, National Cancer Center, Goyang, 10408, Republic of Korea
- Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, 10408, Republic of Korea
| | - Tae Hyun Park
- Department of Anesthesiology, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Kyeong Lee
- College of Pharmacy, Dongguk University-Seoul, Goyang, 10326, Republic of Korea
| | - Byung Il Lee
- Reasearch Institute, National Cancer Center, Goyang, 10408, Republic of Korea.
- Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, 10408, Republic of Korea.
| | - Hyonchol Jang
- Reasearch Institute, National Cancer Center, Goyang, 10408, Republic of Korea.
- Department of Cancer Biomedical Science, National Cancer Center Graduate School of Cancer Science and Policy, Goyang, 10408, Republic of Korea.
| |
Collapse
|
7
|
Zhan Z, Dai F, Zhang T, Chen Y, She J, Jiang H, Liu S, Gu T, Tang L. Oridonin alleviates hyperbilirubinemia through activating LXRα-UGT1A1 axis. Pharmacol Res 2022; 178:106188. [PMID: 35338002 DOI: 10.1016/j.phrs.2022.106188] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 03/14/2022] [Accepted: 03/17/2022] [Indexed: 10/18/2022]
Abstract
Hyperbilirubinemia is a serious hazard to human health due to its neurotoxicity and lethality. So far, successful therapy for hyperbilirubinemia with fewer side effects is still lacking. In this study, we aimed to clarify the effects of oridonin (Ori), an active diterpenoid extracted from Rabdosia rubescens, on hyperbilirubinemia and revealed the underlying molecular mechanism in vivo and in vitro. Here, we showed that liver X receptor alpha (LXRα) deletion eliminated the protective effect of Ori on phenylhydrazine hydrochloride-induced hyperbilirubinemia mice, indicating that LXRα acted as a key target for Ori treatment of hyperbilirubinemia. Ori significantly increased the expression of LXRα and UDP-glucuronosyltransferase 1A1 (UGT1A1) in the liver of wild-type (WT) mice, which were lost in LXRα-/- mice. Ori or LXR agonist GW3965 also reduced lipopolysaccharide/D-galactosamine-induced hyperbilirubinemia via activating LXRα/UGT1A1 in WT mice. Liver UGT1A1 enzyme activity was elevated by Ori or GW3965 in WT mice. Further, Ori up-regulated LXRα gene expression, increased its nuclear translocation and stimulated UGT1A1 promoter activity in HepG2 cells. After silencing LXRα by siRNA, Ori-induced UGT1A1 expression was markedly reduced in HepG2 cells and primary mouse hepatocytes. Taken together, Ori stimulated the transcriptional activity of LXRα, resulting in the up-regulation of UGT1A1. Therefore, Ori or its analogs might have the potential to treat hyperbilirubinemia-related diseases through modulating LXRα-UGT1A1 signaling.
Collapse
Affiliation(s)
- Zhikun Zhan
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Fahong Dai
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Tao Zhang
- Department of Pharmaceutical, Guangzhou Women and Children's Medical Center, Guangzhou, Guangdong 510623, China
| | - Yulian Chen
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Jianglian She
- CAS Key Laboratory of Tropical Marine Bioresources and Ecology, Guangdong Key Laboratory of Marine Materia Medica, South China Sea Institute of Oceanology, Chinese Academy of Sciences, Guangzhou 510301, China
| | - Huanguo Jiang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shuwen Liu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou 510515, China
| | - Tanwei Gu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Lan Tang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China; State Key Laboratory of Organ Failure Research, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
8
|
Mijares MR, Martínez GP, De Sanctis JB. Kauranes as Anti-inflammatory and Immunomodulatory Agents: An Overview of In Vitro and In Vivo Effects. PLANT SECONDARY METABOLITES 2022:191-239. [DOI: 10.1007/978-981-16-4779-6_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
9
|
Targeted theranostic photoactivation on atherosclerosis. J Nanobiotechnology 2021; 19:338. [PMID: 34689768 PMCID: PMC8543964 DOI: 10.1186/s12951-021-01084-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 10/11/2021] [Indexed: 01/08/2023] Open
Abstract
Background Photoactivation targeting macrophages has emerged as a therapeutic strategy for atherosclerosis, but limited targetable ability of photosensitizers to the lesions hinders its applications. Moreover, the molecular mechanistic insight to its phototherapeutic effects on atheroma is still lacking. Herein, we developed a macrophage targetable near-infrared fluorescence (NIRF) emitting phototheranostic agent by conjugating dextran sulfate (DS) to chlorin e6 (Ce6) and estimated its phototherapeutic feasibility in murine atheroma. Also, the phototherapeutic mechanisms of DS-Ce6 on atherosclerosis were investigated. Results The phototheranostic agent DS-Ce6 efficiently internalized into the activated macrophages and foam cells via scavenger receptor-A (SR-A) mediated endocytosis. Customized serial optical imaging-guided photoactivation of DS-Ce6 by light illumination reduced both atheroma burden and inflammation in murine models. Immuno-fluorescence and -histochemical analyses revealed that the photoactivation of DS-Ce6 produced a prominent increase in macrophage-associated apoptotic bodies 1 week after laser irradiation and induced autophagy with Mer tyrosine-protein kinase expression as early as day 1, indicative of an enhanced efferocytosis in atheroma. Conclusion Imaging-guided DS-Ce6 photoactivation was able to in vivo detect inflammatory activity in atheroma as well as to simultaneously reduce both plaque burden and inflammation by harmonic contribution of apoptosis, autophagy, and lesional efferocytosis. These results suggest that macrophage targetable phototheranostic nanoagents will be a promising theranostic strategy for high-risk atheroma. Graphical abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-021-01084-z.
Collapse
|
10
|
Abstract
Terpenoids are the largest class of natural products, most of which are derived from plants. Amongst their numerous biological properties, their anti-tumor effects are of interest for they are extremely diverse which include anti-proliferative, apoptotic, anti-angiogenic, and anti-metastatic activities. Recently, several in vitro and in vivo studies have been dedicated to understanding the 'terpenoid induced autophagy' phenomenon in cancer cells. Light has already been shed on the intricacy of apoptosis and autophagy relationship. This latter crosstalk is driven by the delicate balance between activating or silencing of certain proteins whereby the outcome is expressed via interrelated signaling pathways. In this review, we focus on nine of the most studied terpenoids and on their cell death and autophagic activity. These terpenoids are grouped in three classes: sesquiterpenoid (artemisinin, parthenolide), diterpenoids (oridonin, triptolide), and triterpenoids (alisol, betulinic acid, oleanolic acid, platycodin D, and ursolic acid). We have selected these nine terpenoids among others as they belong to the different major classes of terpenoids and our extensive search of the literature indicated that they were the most studied in terms of autophagy in cancer. These terpenoids alone demonstrate the complexity by which these secondary metabolites induce autophagy via complex signaling pathways such as MAPK/ERK/JNK, PI3K/AKT/mTOR, AMPK, NF-kB, and reactive oxygen species. Moreover, induction of autophagy can be either destructive or protective in tumor cells. Nevertheless, should this phenomenon be well understood, we ought to be able to exploit it to create novel therapies and design more effective regimens in the management and treatment of cancer.
Collapse
|
11
|
Kang N, Cao S, Jiang B, Zhang Q, Donkor PO, Zhu Y, Qiu F, Gao X. Cetuximab enhances oridonin-induced apoptosis through mitochondrial pathway and endoplasmic reticulum stress in laryngeal squamous cell carcinoma cells. Toxicol In Vitro 2020; 67:104885. [PMID: 32407876 DOI: 10.1016/j.tiv.2020.104885] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 05/08/2020] [Accepted: 05/09/2020] [Indexed: 12/24/2022]
Abstract
Cetuximab plus oridonin showed a synergistic way to kill laryngeal squamous cell carcinoma (LSCC), as been reported previously. The present work further mechanistically extended action of the synergistic effects of combination treatment. Firstly, two LSCC cells displayed higher sensitivity to oridonin, whereas both low EGFR expression tumor cells and EGFR knockdown LSCC cells were less sensitive to oridonin. Next, cetuximab/oridonin significantly enhanced the mitochondrial apoptosis through NF-κB. Meanwhile, PI3K/Akt and JAK2/STAT3 pathways are associated with the nucleus translocation of NF-κB by combination treatment. Additionally, cetuximab enhanced oridonin-promoted ER stress-related apoptosis. Interestingly, both ER stress and mitochondrial apoptosis by combination treatment are abrogated by ROS scavenger. Furthermore, oridonin/cetuximab induced ROS production after 1.5 h, followed by G2/M arrest and apoptosis, indicating that ROS generation might be an early and key event. Taken together, cetuximab enhances oridonin-induced ER stress and mitochondrial apoptotic pathway, which contributes to the synergistic antitumor effects of cetuximab/oridonin.
Collapse
Affiliation(s)
- Ning Kang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Shijie Cao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Benke Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Faculty of Life Sciences and Biological Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Qiang Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Paul Owusu Donkor
- School of Pharmacy, University of Ghana, Korle Bu, Accra, P.O. Box 52, Ghana
| | - Yan Zhu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Feng Qiu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| | - Xiumei Gao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| |
Collapse
|
12
|
Lee HA, Park MH, Song Y, Na HS, Chung J. Role of
Aggregatibacter actinomycetemcomitans‐
induced autophagy in inflammatory response. J Periodontol 2020; 91:1682-1693. [DOI: 10.1002/jper.19-0639] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 02/13/2020] [Accepted: 02/16/2020] [Indexed: 01/26/2023]
Affiliation(s)
- Hyun Ah Lee
- Department of Oral Microbiology School of Dentistry Pusan National University Yangsan Korea
| | - Mi Hee Park
- Department of Oral Microbiology School of Dentistry Pusan National University Yangsan Korea
- Oral Genomics Research Center Pusan National University Yangsan Korea
| | - Yuri Song
- Department of Oral Microbiology School of Dentistry Pusan National University Yangsan Korea
- Oral Genomics Research Center Pusan National University Yangsan Korea
| | - Hee Sam Na
- Department of Oral Microbiology School of Dentistry Pusan National University Yangsan Korea
- Oral Genomics Research Center Pusan National University Yangsan Korea
| | - Jin Chung
- Department of Oral Microbiology School of Dentistry Pusan National University Yangsan Korea
- Oral Genomics Research Center Pusan National University Yangsan Korea
| |
Collapse
|
13
|
Cao S, Huang Y, Zhang Q, Lu F, Donkor PO, Zhu Y, Qiu F, Kang N. Molecular mechanisms of apoptosis and autophagy elicited by combined treatment with oridonin and cetuximab in laryngeal squamous cell carcinoma. Apoptosis 2020; 24:33-45. [PMID: 30430397 DOI: 10.1007/s10495-018-1497-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Combined oridonin (ORI), a natural and safe kaurene diterpenoid isolated from Rabdosia rubescens, and cetuximab (Cet), an anti-EGFR monoclonal antibody, have been reported to exert synergistic anti-tumor effects against laryngeal squamous cell carcinoma (LSCC) both in vitro and in vivo by our group. In the present study, we further found that ORI/Cet treatment not only resulted in apoptosis but also induced autophagy. AMPK/mTOR signaling pathway was found to be involved in the activation of autophagy in ORI/Cet-treated LSCC cells, which is independent of p53 status. Additionally, chromatin immunoprecipitation (ChIP) assay showed that ORI/Cet significantly increased the binding NF-κB family member p65 with the promotor of BECN 1, and p65-mediated up-regulation of BECN 1 caused by ORI/Cet is coupled to increased autophagy. On the other hand, we demonstrated that either Beclin 1 SiRNA or autophagy inhibitors could increase ORI/Cet induced-apoptosis, indicating that autophagy induced by combination of the two agents plays a cytoprotective role. Interestingly, 48 h after the combined treatment, autophagy began to decrease but apoptosis was significantly elevated. Our findings suggest that autophagy might be strongly associated with the antitumor efficacy of ORI/Cet, which may be beneficial to the clinical application of ORI/Cet in LSCC treatment.
Collapse
Affiliation(s)
- Shijie Cao
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, People's Republic of China
| | - Yiyuan Huang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, 312 Anshanxi Road, Tianjin, 300193, People's Republic of China.,Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, People's Republic of China
| | - Qiang Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, 312 Anshanxi Road, Tianjin, 300193, People's Republic of China
| | - Fangjin Lu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, People's Republic of China
| | - Paul Owusu Donkor
- School of Pharmacy, University of Health and Allied Sciences, Ho, PMB 31, Ghana
| | - Yan Zhu
- Tianjin State Key Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, People's Republic of China
| | - Feng Qiu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, People's Republic of China
| | - Ning Kang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, 312 Anshanxi Road, Tianjin, 300193, People's Republic of China.
| |
Collapse
|
14
|
Wan LQ, Tan Y, Jiang M, Hua Q. The prognostic impact of traditional Chinese medicine monomers on tumor-associated macrophages in non-small cell lung cancer. Chin J Nat Med 2020; 17:729-737. [PMID: 31703753 DOI: 10.1016/s1875-5364(19)30089-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Indexed: 02/06/2023]
Abstract
Non-small cell lung cancer (NSCLC) accounts for 80%-85% of all lung malignancies and good diagnosis and prognosis of NSCLC are critical to the increase of its survival rate. Tumor-associated macrophages (TAM) abundantly present in numerous cancer types, and the role of TAMs in tumor biology and their prognostic value in cancer become major topics of interest. After various stimulations in the tumor microenvironment, TAMs develop into a M1 (tumor-inhibitory) phenotype or M2 (tumor-promoting) phenotype. Recent studies show that traditional Chinese medicine (TCM) monomers have markedly inhibitory actions for NSCLC through M1/M2 modulation. Due to the TCM monomers mainly covered five categories, i.e. terpenoids, flavonoids, polysaccharides, natural polyphenols, and alkaloids. Thus, we will discuss the regulation of TCM monomers on TAM involve in these five parts in this review. In addition, the potential role of TAMs as therapeutic targets will be discussed.
Collapse
Affiliation(s)
- Liang-Qin Wan
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Yan Tan
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Miao Jiang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Qian Hua
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
15
|
Zhao YL, Zhang X, Liu WW, Yang YT, Gao ZK, Liu XL, Liu W, Hayashi T, Yamato M, Fujisaki H, Hattori S, Mizuno K, Atsuzawa Y, Tashiro SI, Onodera S, Ikejima T. Reactive oxygen species are responsible for the cell aggregation and production of pro-inflammatory mediators in phorbol ester (PMA)-treated U937 cells on gelatin-coated dishes through upregulation of autophagy. Connect Tissue Res 2019; 60:323-334. [PMID: 30277081 DOI: 10.1080/03008207.2018.1530770] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Purpose: Our previous studies indicate that phorbol 12-myristate 13-acetate (PMA)-treated U937 cells cultured on collagen I-coated dishes express lowered production of pro-inflammatory mediators in parallel through reduced reactive oxygen species (ROS) levels. By contrast, PMA-treated U937 cells on gelatin, the denatured collagen, show enhanced production of pro-inflammatory mediators, mediated by up-regulating autophagy levels. The present study is aimed to investigate the effect of ROS levels in PMA-treated U937 cells cultured on gelatin-coated surface. Material and methods: MTT assay, flow cytometric analysis of ROS and autophagy, biochemical detection of antioxidant levels, enzyme-linked immunosorbent assay, and western blot were used. Results: Gelatin-coating increased ROS levels in PMA-treated U937 cells. Increased ROS levels are involved in the regulation of cell aggregation and the release of pro-inflammatory mediators in gelatin-coated culture. These results lead to the query about the crosstalk between the two positive regulators, the autophagy and ROS. Autophagy induction is attenuated by N-acetyl-L-cysteine treatment, but the treatment with autophagy inhibitor, 3-methyladenine, does not affect ROS levels, suggesting ROS are upstream of autophagy in the regulation axis of differentiated U937 cells on gelatin-coated surface. Further study confirmed that upregulation of autophagy was responsible for ROS-induced cell aggregation and production of pro-inflammatory mediators. Conclusion: The results suggest that gelatin-coating promotes the aggregation of PMA-treated U937 cells and the production of pro-inflammatory mediators by ROS-autophagy signaling pathway.
Collapse
Affiliation(s)
- Ye-Li Zhao
- a China-Japan Research Institute of Medical and Pharmaceutical Sciences , Shenyang Pharmaceutical University , Shenyang , China
| | - Xuan Zhang
- a China-Japan Research Institute of Medical and Pharmaceutical Sciences , Shenyang Pharmaceutical University , Shenyang , China
| | - Wei-Wei Liu
- a China-Japan Research Institute of Medical and Pharmaceutical Sciences , Shenyang Pharmaceutical University , Shenyang , China
| | - Yu-Ting Yang
- a China-Japan Research Institute of Medical and Pharmaceutical Sciences , Shenyang Pharmaceutical University , Shenyang , China
| | - Zhuo-Kun Gao
- a China-Japan Research Institute of Medical and Pharmaceutical Sciences , Shenyang Pharmaceutical University , Shenyang , China
| | - Xiao-Ling Liu
- a China-Japan Research Institute of Medical and Pharmaceutical Sciences , Shenyang Pharmaceutical University , Shenyang , China
| | - Wei Liu
- a China-Japan Research Institute of Medical and Pharmaceutical Sciences , Shenyang Pharmaceutical University , Shenyang , China
| | - Toshihiko Hayashi
- a China-Japan Research Institute of Medical and Pharmaceutical Sciences , Shenyang Pharmaceutical University , Shenyang , China
| | - Masayuki Yamato
- b Institute of Advanced Biomedical Engineering and Science , Tokyo Women's Medical University , Tokyo , Japan
| | - Hitomi Fujisaki
- c Nippi Research Institute of Biomatrix , Toride, Ibaraki , Japan
| | - Shunji Hattori
- c Nippi Research Institute of Biomatrix , Toride, Ibaraki , Japan
| | - Kazunori Mizuno
- c Nippi Research Institute of Biomatrix , Toride, Ibaraki , Japan
| | - Yuji Atsuzawa
- c Nippi Research Institute of Biomatrix , Toride, Ibaraki , Japan
| | - Shin-Ichi Tashiro
- d Department of Medical Education and Primary Care , Kyoto Prefectural University of Medicine , Kyoto , Japan
| | - Satoshi Onodera
- e Department of Clinical and Pharmaceutical Sciences , Showa Pharmaceutical University , Tokyo , Japan
| | - Takashi Ikejima
- a China-Japan Research Institute of Medical and Pharmaceutical Sciences , Shenyang Pharmaceutical University , Shenyang , China
| |
Collapse
|
16
|
Natural Compound Oridonin Inhibits Endotoxin-Induced Inflammatory Response of Activated Hepatic Stellate Cells. BIOMED RESEARCH INTERNATIONAL 2018; 2018:6137420. [PMID: 30687752 PMCID: PMC6330820 DOI: 10.1155/2018/6137420] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 09/16/2018] [Accepted: 12/09/2018] [Indexed: 12/17/2022]
Abstract
Hepatic stellate cells (HSCs) play an important role in hepatic fibrogenesis and inflammatory modulation. Endotoxin is dramatically increased in portal venous blood after serious injury and can contribute to liver damage. However, the mechanism underlying endotoxin's effects on HSCs remains largely unknown. Oridonin is a bioactive diterpenoid isolated from Rabdosia rubescens that exhibits anti-inflammatory properties in different tissues. In the present study, we determined the effects of oridonin on endotoxin-induced inflammatory response and signaling pathways in vitro. The production of proinflammatory cytokines in activated human HSCs line LX-2 was measured by ELISA and Western blots. Immunofluorescence and nuclear fractionation assay were used to determine NF-κB activity. Oridonin treatment significantly inhibited LPS-induced proinflammatory cytokines IL-1β, IL-6, and MCP-1 production as well as cell adhesion molecules ICAM-1 and VCAM-1. Additionally, oridonin blocked LPS-induced NF-κB p65 nuclear translocation and DNA binding activity. Oridonin prevented LPS-stimulated NF-κB regulator IKKα/β and IκBα phosphorylation and IκBα degradation. Combined treatment of oridonin and an Hsp70 substrate binding inhibitor synergistically suppressed LPS-stimulated proinflammatory cytokines and NF-κB pathway activation. Therefore, oridonin inhibits LPS-stimulated proinflammatory mediators through IKK/IκBα/NF-κB pathway. Oridonin could be a promising agent for a hepatic anti-inflammatory.
Collapse
|
17
|
Chen YD, Fang YT, Cheng YL, Lin CF, Hsu LJ, Wang SY, Anderson R, Chang CP, Lin YS. Exophagy of annexin A2 via RAB11, RAB8A and RAB27A in IFN-γ-stimulated lung epithelial cells. Sci Rep 2017; 7:5676. [PMID: 28720835 PMCID: PMC5516008 DOI: 10.1038/s41598-017-06076-4] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 06/07/2017] [Indexed: 12/09/2022] Open
Abstract
Annexin A2 (ANXA2), a phospholipid-binding protein, has multiple biological functions depending on its cellular localization. We previously demonstrated that IFN-γ-triggered ANXA2 secretion is associated with exosomal release. Here, we show that IFN-γ-induced autophagy is essential for the extracellular secretion of ANXA2 in lung epithelial cells. We observed colocalization of ANXA2-containing autophagosomes with multivesicular bodies (MVBs) after IFN-γ stimulation, followed by exosomal release. IFN-γ-induced exophagic release of ANXA2 could not be observed in ATG5-silenced or mutant RAB11-expressing cells. Furthermore, knockdown of RAB8A and RAB27A, but not RAB27B, reduced IFN-γ-triggered ANXA2 secretion. Surface translocation of ANXA2 enhanced efferocytosis by epithelial cells, and inhibition of different exophagic steps, including autophagosome formation, fusion of autophagosomes with MVBs, and fusion of amphisomes with plasma membrane, reduced ANXA2-mediated efferocytosis. Our data reveal a novel route of IFN-γ-induced exophagy of ANXA2.
Collapse
Affiliation(s)
- Ying-Da Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Ting Fang
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Lin Cheng
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chiou-Feng Lin
- Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Microbiology and Immunology, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Li-Jin Hsu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shu-Ying Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Robert Anderson
- Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan.,Departments of Microbiology & Immunology and Pediatrics, and Canadian Center for Vaccinology, Dalhousie University, Halifax, Canada
| | - Chih-Peng Chang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan. .,Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan. .,Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Yee-Shin Lin
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan. .,Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan, Taiwan. .,Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
18
|
Zhang Y, Liang Y, He C. Anticancer activities and mechanisms of heat-clearing and detoxicating traditional Chinese herbal medicine. Chin Med 2017; 12:20. [PMID: 28702078 PMCID: PMC5506596 DOI: 10.1186/s13020-017-0140-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/30/2017] [Indexed: 02/07/2023] Open
Abstract
In traditional Chinese medicine (TCM) theory, pathogenic heat and toxins, which are akin to the inflammatory factors, are the causes of cancer and could promote its virulent development. Therefore, heat-clearing and detoxicating (HCD) herbs are essential components of TCM formulas for cancer treatment. An increasing interest has been focused on the study of HCD herbs and accumulated evidences have shown that HCD herbs or HCD herbs-based formulas exhibited remarkable anticancer effects when used alone or combined with other therapeutic approaches. Some of the HCD herb-derived products have been tested in clinical trials. Studies revealed that extracts or pure compounds of the HCD herbs showed a broad anticancer spectrum against both solid and hematologic malignancies without significant toxic effects. Notably, some HCD herbs or formulas could strongly enhance the anticancer activities of chemo- or radio-therapy and alleviate their side effects. The anticancer activities of HCD herb exacts or the pure compounds were reported to be through multiple cellular or molecular mechanisms, such as induction of cancer cell apoptosis, differentiation and cell cycle arrest, inhibition of cancer cell growth, invasion and metastasis, and inhibition of tumor angiogenesis. In this review, we provide comprehensive analysis and summary of research progress and future prospects in this field to facilitate the further study and application of HCD herbs.
Collapse
Affiliation(s)
- Yulin Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, N22-7038, Avenida da Universidade, Taipa, Macao, 999078 China
| | - Yeer Liang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, N22-7038, Avenida da Universidade, Taipa, Macao, 999078 China
| | - Chengwei He
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, N22-7038, Avenida da Universidade, Taipa, Macao, 999078 China
| |
Collapse
|
19
|
Spirin P, Lebedev T, Orlova N, Morozov A, Poymenova N, Dmitriev SE, Buzdin A, Stocking C, Kovalchuk O, Prassolov V. Synergistic suppression of t(8;21)-positive leukemia cell growth by combining oridonin and MAPK1/ERK2 inhibitors. Oncotarget 2017; 8:56991-57002. [PMID: 28915648 PMCID: PMC5593619 DOI: 10.18632/oncotarget.18503] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 04/18/2017] [Indexed: 01/03/2023] Open
Abstract
One of the most common chromosomal translocations in acute myeloid leukemia is t(8;21)(q22;q22), which results in the appearance of abnormal transcripts encoding for the fusion protein RUNX1-ETO. Therefore, this oncoprotein is considered to be a pertinent and promising target for treating t(8;21) leukemia. Previously, we have shown that downregulation of RUNX1-ETO leads to activation of intracellular signaling pathways enhancing cell survival and determined that the protein ERK2 can mediate activation of most of these pathways. Here we used a combination of oridonin (natural tetracycline diterpenoid), which has been shown to exhibit anti-RUNX1-ETO activity, and ERK2 kinase inhibitors. We found that treatment of leukemic t(8;21)-positive Kasumi-1 cells with oridonin cause decrease of phosphorylated ERK1/2. Treatment of these cells with ERK2 inhibitors makes them more sensitive to RUNX1-ETO inhibition with oridonin. Therefore we postulate that simultaneous inhibition of RUNX1-ETO and ERK2 cause synergistic effect on survival of leukemic cells.
Collapse
Affiliation(s)
- Pavel Spirin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia
| | - Timofey Lebedev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia
| | - Natalia Orlova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia
| | - Alexey Morozov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia
| | - Nadezhda Poymenova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia
| | - Sergey E Dmitriev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia.,Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Anton Buzdin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia.,Dmitry Rogachev Federal Research Center of Pediatric Hematology, Oncology and Immunology, Moscow 117997, Russia.,National Research Centre "Kurchatov Institute", Centre for Convergence of Nano-, Bio-, Information and Cognitive Sciences and Technologies, Moscow 123182, Russia
| | - Carol Stocking
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Olga Kovalchuk
- OncoFinder Ltd, Lethbridge, AB T1K7×8, Canada.,Department of Biological Sciences, University of Lethbridge, Lethbridge, AB T1K3M4, Canada
| | - Vladimir Prassolov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow 119991, Russia
| |
Collapse
|
20
|
Ouimet M, Ediriweera H, Afonso MS, Ramkhelawon B, Singaravelu R, Liao X, Bandler RC, Rahman K, Fisher EA, Rayner KJ, Pezacki JP, Tabas I, Moore KJ. microRNA-33 Regulates Macrophage Autophagy in Atherosclerosis. Arterioscler Thromb Vasc Biol 2017; 37:1058-1067. [PMID: 28428217 DOI: 10.1161/atvbaha.116.308916] [Citation(s) in RCA: 167] [Impact Index Per Article: 20.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2016] [Accepted: 04/05/2017] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Defective autophagy in macrophages leads to pathological processes that contribute to atherosclerosis, including impaired cholesterol metabolism and defective efferocytosis. Autophagy promotes the degradation of cytoplasmic components in lysosomes and plays a key role in the catabolism of stored lipids to maintain cellular homeostasis. microRNA-33 (miR-33) is a post-transcriptional regulator of genes involved in cholesterol homeostasis, yet the complete mechanisms by which miR-33 controls lipid metabolism are unknown. We investigated whether miR-33 targeting of autophagy contributes to its regulation of cholesterol homeostasis and atherogenesis. APPROACH AND RESULTS Using coherent anti-Stokes Raman scattering microscopy, we show that miR-33 drives lipid droplet accumulation in macrophages, suggesting decreased lipolysis. Inhibition of neutral and lysosomal hydrolysis pathways revealed that miR-33 reduced cholesterol mobilization by a lysosomal-dependent mechanism, implicating repression of autophagy. Indeed, we show that miR-33 targets key autophagy regulators and effectors in macrophages to reduce lipid droplet catabolism, an essential process to generate free cholesterol for efflux. Notably, miR-33 regulation of autophagy lies upstream of its known effects on ABCA1 (ATP-binding cassette transporter A1)-dependent cholesterol efflux, as miR-33 inhibitors fail to increase efflux upon genetic or chemical inhibition of autophagy. Furthermore, we find that miR-33 inhibits apoptotic cell clearance via an autophagy-dependent mechanism. Macrophages treated with anti-miR-33 show increased efferocytosis, lysosomal biogenesis, and degradation of apoptotic material. Finally, we show that treating atherosclerotic Ldlr-/- mice with anti-miR-33 restores defective autophagy in macrophage foam cells and plaques and promotes apoptotic cell clearance to reduce plaque necrosis. CONCLUSIONS Collectively, these data provide insight into the mechanisms by which miR-33 regulates cellular cholesterol homeostasis and atherosclerosis.
Collapse
Affiliation(s)
- Mireille Ouimet
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Hasini Ediriweera
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Milessa Silva Afonso
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Bhama Ramkhelawon
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Ragunath Singaravelu
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Xianghai Liao
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Rachel C Bandler
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Karishma Rahman
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Edward A Fisher
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Katey J Rayner
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - John P Pezacki
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Ira Tabas
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.)
| | - Kathryn J Moore
- From the Marc and Ruti Bell Vascular Biology and Disease Program, Leon H. Charney Division of Cardiology, Department of Medicine (M.O., H.E., M.S.A., R.C.B., K.R., E.A.F., K.J.M.) and Division of Vascular Surgery, Department of Surgery (B.R.), New York University Medical Center; Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Canada (R.S., K.J.R., J.P.P.); National Research Council of Canada, Ottawa, Ontario (R.S., J.P.P.); Departments of Medicine, Pathology and Cell Biology, Columbia University, New York (X.L., I.T.); and University of Ottawa Heart Institute, Ontario, Canada (K.J.R.).
| |
Collapse
|
21
|
Chen YD, Fang YT, Chang CP, Lin CF, Hsu LJ, Wu SR, Chiu YC, Anderson R, Lin YS. S100A10 Regulates ULK1 Localization to ER-Mitochondria Contact Sites in IFN-γ-Triggered Autophagy. J Mol Biol 2016; 429:142-157. [PMID: 27871932 DOI: 10.1016/j.jmb.2016.11.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2016] [Revised: 10/28/2016] [Accepted: 11/15/2016] [Indexed: 01/07/2023]
Abstract
During the process of autophagy, the autophagy-related proteins are translocated to autophagosome formation sites. Here, we demonstrate that S100A10 is required for ULK1 localization to autophagosome formation sites. Silencing of S100A10 reduces IFN-γ-induced autophagosome formation. We also determined the role of annexin A2 (ANXA2), a binding partner of S100A10, which has been reported to promote phagophore assembly. Silencing of ANXA2 reduced S100A10 expression. However, overexpression of S100A10 in ANXA2-silenced cells was still able to enhance autophagosome formation, suggesting that ANXA2 regulates IFN-γ-induced autophagy through S100A10. We also observed that S100A10 interacted with ULK1 after IFN-γ stimulation, and S100A10 knockdown prevented ULK1 localization to autophagosome formation sites. Finally, the release of high mobility group protein B1, one of the functions mediated by IFN-γ-induced autophagy, was inhibited in S100A10 knockdown cells. These results elucidate the importance of S100A10 in autophagosome formation and reveal the relationship between S100A10 and ULK1 in IFN-γ-induced autophagy.
Collapse
Affiliation(s)
- Ying-Da Chen
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Yi-Ting Fang
- Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Chih-Peng Chang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Chiou-Feng Lin
- Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; Department of Microbiology and Immunology, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Li-Jin Hsu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Shang-Rung Wu
- Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Yen-Chi Chiu
- Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan
| | - Robert Anderson
- Departments of Microbiology & Immunology and Pediatrics, and Canadian Center for Vaccinology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Yee-Shin Lin
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; Department of Microbiology and Immunology, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan; Center of Infectious Disease and Signaling Research, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan.
| |
Collapse
|
22
|
Zhou P, Tan YZ, Wang HJ, Li T, He T, Yu Y, Zhang J, Zhang D. Cytoprotective effect of autophagy on phagocytosis of apoptotic cells by macrophages. Exp Cell Res 2016; 348:165-176. [PMID: 27658567 DOI: 10.1016/j.yexcr.2016.09.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 09/18/2016] [Indexed: 12/29/2022]
Abstract
Clearance of the apoptotic cells by phagocytes plays pivotal roles in maintenance of tissue homeostasis, promotion of immunological tolerance and anti-inflammatory response. Recent studies show that autophagy is involved in phagocytosis of the apoptotic cells. However, contribution of autophagy to phagocytosis of the apoptotic cells by macrophages is not clearly defined. Here, we assessed cytoprotective effect of autophagy on clearance of the apoptotic cells. Apoptosis of murine splenic lymphocytes and human T-cell leukemia cells was induced with cyclophosphamide. After engulfment of the apoptotic cells, expression of Belin-1 and LC3 in macrophages was upregulated, the number of MDC-positive vesicles, LC3-positive autophagosomes and autophagic ultrastructures increased significantly. Autophagosome was fused with phagosome containing fragments of the nuclei or other debris of the apoptotic cells to form amphisome. Some cells in macrophages phagocytosing the apoptotic cells became apoptotic. After autophagy of macrophages was inhibited with 3-MA, viability and survival of macrophages reduced, phagocytosis of the apoptotic cells by macrophages deceased significantly. These results demonstrate that autophagy plays an important role in promoting clearance of the apoptotic cells by protecting macrophages from apoptosis during phagocytosis as well as degrading the contents of phagosomes via amphisome formation.
Collapse
Affiliation(s)
- Pei Zhou
- Department of Anatomy, Histology and Embryology, Shanghai Medical School of Fudan University, Shanghai 200032, China
| | - Yu-Zhen Tan
- Department of Anatomy, Histology and Embryology, Shanghai Medical School of Fudan University, Shanghai 200032, China
| | - Hai-Jie Wang
- Department of Anatomy, Histology and Embryology, Shanghai Medical School of Fudan University, Shanghai 200032, China.
| | - Ting Li
- Department of Anatomy, Histology and Embryology, Shanghai Medical School of Fudan University, Shanghai 200032, China
| | - Tao He
- Department of Anatomy, Histology and Embryology, Shanghai Medical School of Fudan University, Shanghai 200032, China
| | - Ying Yu
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Jian Zhang
- Key Laboratory of Food Safety Research, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| | - Dan Zhang
- Department of Anatomy, Histology and Embryology, Shanghai Medical School of Fudan University, Shanghai 200032, China
| |
Collapse
|
23
|
Activation of Adenosine 2A receptor inhibits neutrophil apoptosis in an autophagy-dependent manner in mice with systemic inflammatory response syndrome. Sci Rep 2016; 6:33614. [PMID: 27647162 PMCID: PMC5028892 DOI: 10.1038/srep33614] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 08/30/2016] [Indexed: 01/13/2023] Open
Abstract
Systemic inflammatory response syndrome (SIRS) is an overwhelming whole body inflammation caused by infectious diseases or sterile insults. Neutrophils are the dominant participants during inflammation, and their survival and death determine the initiation as well as resolution of SIRS. Apoptosis and autophagy are two fundamental cellular processes that modulating cell fate, but their correlation and regulators in neutrophils under SIRS condition have not been elucidated. In this study, we demonstrated that high dose of LPS induced both apoptosis and autophagy of neutrophils in a mouse SIRS model and LPS-stimulated neutrophils in vitro. Moreover, we found that the adenosine 2A receptor (A2AR), a known anti-inflammatory G protein-coupled receptor (GPCR), could inhibit LPS-induced neutrophil apoptosis by suppressing the LPS-induced autophagy. Activation of A2AR suppressed LPS-induced autophagy by inhibiting the ROS-JNK pathway as well as promoting GPCR βϒ subunit–AKT signaling. The A2AR-inhibited autophagy suppressed apoptosis of neutrophils by blocking caspase8, caspase3 and PARP signaling. These findings not only increase our understandings of neutrophils’ fate and function in response to systemic inflammation, but also identify a novel anti-inflammatory role of A2AR in modulating neutrophils’ survival during inflammation.
Collapse
|
24
|
Wang C, Jiang L, Wang S, Shi H, Wang J, Wang R, Li Y, Dou Y, Liu Y, Hou G, Ke Y, Liu H. The Antitumor Activity of the Novel Compound Jesridonin on Human Esophageal Carcinoma Cells. PLoS One 2015; 10:e0130284. [PMID: 26103161 PMCID: PMC4477902 DOI: 10.1371/journal.pone.0130284] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2014] [Accepted: 05/18/2015] [Indexed: 11/18/2022] Open
Abstract
Jesridonin, a small molecule obtained through the structural modification of Oridonin, has extensive antitumor activity. In this study, we evaluated both its in vitro activity in the cancer cell line EC109 and its in vivo effect on tumor xenografts in nude mice. Apoptosis induced by Jesridonin was determined using an MTT assay, Annexin-V FITC assay and Hoechest 33258 staining. Apoptosis via mitochondrial and death receptor pathways were confirmed by detecting the regulation of MDM2, p53, and Bcl-2 family members and by activation of caspase-3/-8/-9. In addition, vena caudalis injection of Jesridonin showed significant inhibition of tumor growth in the xenograft model, and Jesridonin-induced cell apoptosis in tumor tissues was determined using TUNEL. Biochemical serum analysis of alkaline phosphatase (ALP), alanine transaminase (ALT), aspartate transaminase (AST), gamma-glutamyl transferase (GGT), total protein (TP) and albumin (ALB) indicated no obvious effects on liver function. Histopathological examination of the liver, kidney, lung, heart and spleen revealed no signs of JD-induced toxicity. Taken together, these results demonstrated that Jesridonin exhibits antitumor activity in human esophageal carcinomas EC109 cells both in vitro and in vivo and demonstrated no adverse effects on major organs in nude mice. These studies provide support for new drug development.
Collapse
Affiliation(s)
- Cong Wang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Liping Jiang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Saiqi Wang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Hongge Shi
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Junwei Wang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Ran Wang
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Yongmei Li
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Yinhui Dou
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Ying Liu
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Guiqin Hou
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
| | - Yu Ke
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- * E-mail: (HML); (YK)
| | - Hongmin Liu
- School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- New Drug Research & Development Center, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan, 450001, China
- * E-mail: (HML); (YK)
| |
Collapse
|
25
|
Li X, Yang Z. Interaction of oridonin with human serum albumin by isothermal titration calorimetry and spectroscopic techniques. Chem Biol Interact 2015; 232:77-84. [PMID: 25816984 DOI: 10.1016/j.cbi.2015.03.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Revised: 03/09/2015] [Accepted: 03/17/2015] [Indexed: 11/19/2022]
Abstract
Oridonin has been traditionally and widely used for treatment of various human diseases due to its uniquely biological, pharmacological and physiological functions. In this study, the interaction between oridonin and human serum albumin (HSA) was investigated using isothermal titration calorimetry (ITC), in combination with fluorescence spectroscopy and UV-vis absorption spectroscopy. We found that the hydrogen bond and van der Waals force are the major binding forces in the binding of oridonin to HSA. The binding of oridonin to HSA is driven by favorable enthalpy and unfavorable entropy. Oridonin can quench the fluorescence of HSA through a static quenching mechanism. The binding constant between oridonin and HSA is moderate and the equilibrium fraction of unbound oridonin f(u) > 60%. Binding site I is found to be the primary binding site for oridonin. Additionally, oridonin may induce conformational changes of HSA and affect its biological function as the carrier protein. The results of the current study suggest that oridonin can be stored and transported from the circulatory system to reach its target organ to provide its therapeutic effects. But its side-effect in the clinics cannot be overlook. The study provides an accurate and full basic data for clarifying the binding mechanism of oridonin with HSA and is helpful for understanding its effect on protein function during the blood transportation process and its biological activity in vivo.
Collapse
Affiliation(s)
- Xiangrong Li
- Department of Chemistry, School of Basic Medicine, Xinxiang Medical University, Xinxiang, Henan 453003, PR China.
| | - Zhenhua Yang
- Department of Chemistry, School of Basic Medicine, Xinxiang Medical University, Xinxiang, Henan 453003, PR China
| |
Collapse
|
26
|
Schapansky J, Nardozzi JD, LaVoie MJ. The complex relationships between microglia, alpha-synuclein, and LRRK2 in Parkinson's disease. Neuroscience 2014; 302:74-88. [PMID: 25284317 DOI: 10.1016/j.neuroscience.2014.09.049] [Citation(s) in RCA: 93] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 09/22/2014] [Accepted: 09/23/2014] [Indexed: 12/19/2022]
Abstract
The proteins alpha-synuclein (αSyn) and leucine rich repeat kinase 2 (LRRK2) are both key players in the pathogenesis of the neurodegenerative disorder Parkinson's disease (PD), but establishing a functional link between the two proteins has proven elusive. Research studies for these two proteins have traditionally and justifiably focused in neuronal cells, but recent studies indicate that each protein could play a greater pathological role elsewhere. αSyn is expressed at high levels within neurons, but they also secrete the protein into the extracellular milieu, where it can have broad ranging effects in the nervous system and relevance to disease etiology. Similarly, low neuronal LRRK2 expression and activity suggests that LRRK2-related functions could be more relevant in cells with higher expression, such as brain-resident microglia. Microglia are monocytic immune cells that protect neurons from noxious stimuli, including pathological αSyn species, and microglial activation is believed to contribute to neuroinflammation and neuronal death in PD. Interestingly, both αSyn and LRRK2 can be linked to microglial function. Secreted αSyn can directly activate microglia, and can be taken up by microglia for clearance, while LRRK2 has been implicated in the intrinsic regulation of microglial activation and of lysosomal degradation processes. Based on these observations, the present review will focus on how PD-associated mutations in LRRK2 could potentially alter microglial biology with respect to neuronally secreted αSyn, resulting in cell dysfunction and neurodegeneration.
Collapse
Affiliation(s)
- J Schapansky
- Center for Neurologic Diseases, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA 02115, United States
| | - J D Nardozzi
- Center for Neurologic Diseases, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA 02115, United States
| | - M J LaVoie
- Center for Neurologic Diseases, Harvard Medical School, and Brigham and Women's Hospital, Boston, MA 02115, United States.
| |
Collapse
|
27
|
Szondy Z, Garabuczi E, Joós G, Tsay GJ, Sarang Z. Impaired clearance of apoptotic cells in chronic inflammatory diseases: therapeutic implications. Front Immunol 2014; 5:354. [PMID: 25136342 PMCID: PMC4117929 DOI: 10.3389/fimmu.2014.00354] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Accepted: 07/09/2014] [Indexed: 12/14/2022] Open
Abstract
In healthy individuals, billions of cells die by apoptosis every day. Removal of the dead cells by phagocytosis (a process called efferocytosis) must be efficient to prevent secondary necrosis and the consequent release of pro-inflammatory cell contents that damages the tissue environment and provokes autoimmunity. In addition, detection and removal of apoptotic cells generally induces an anti-inflammatory response. As a consequence improper clearance of apoptotic cells, being the result of either genetic anomalies and/or a persistent disease state, contributes to the establishment and progression of a number of human chronic inflammatory diseases such as autoimmune and neurological disorders, inflammatory lung diseases, obesity, type 2 diabetes, or atherosclerosis. During the past decade, our knowledge about the mechanism of efferocytosis has significantly increased, providing therapeutic targets through which impaired phagocytosis of apoptotic cells and the consequent inflammation could be influenced in these diseases.
Collapse
Affiliation(s)
- Zsuzsa Szondy
- Department of Dental Biochemistry, Faculty of Dentistry, University of Debrecen , Debrecen , Hungary
| | - Eva Garabuczi
- Department of Dental Biochemistry, Faculty of Dentistry, University of Debrecen , Debrecen , Hungary
| | - Gergely Joós
- Department of Dental Biochemistry, Faculty of Dentistry, University of Debrecen , Debrecen , Hungary
| | - Gregory J Tsay
- Department of Internal Medicine, Faculty of Medicine, Chung Shan Medical University Hospital , Taichung , Taiwan
| | - Zsolt Sarang
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen , Debrecen , Hungary
| |
Collapse
|
28
|
Fan J, Zeng X, Li Y, Wang S, Wang Z, Sun Y, Gao H, Zhang G, Feng M, Ju D. Autophagy plays a critical role in ChLym-1-induced cytotoxicity of non-hodgkin's lymphoma cells. PLoS One 2013; 8:e72478. [PMID: 24015249 PMCID: PMC3756084 DOI: 10.1371/journal.pone.0072478] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 07/10/2013] [Indexed: 12/15/2022] Open
Abstract
Autophagy is a critical mechanism in both cancer therapy resistance and tumor suppression. Monoclonal antibodies have been documented to kill tumor cells via apoptosis, antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). In this study, we report for the first time that chLym-1, a chimeric anti-human HLA-DR monoclonal antibody, induces autophagy in Raji Non-Hodgkin’s Lymphoma (NHL) cells. Interestingly, inhibition of autophagy by pharmacological inhibitors (3-methyladenine and NH4Cl) or genetic approaches (siRNA targeting Atg5) suppresses chLym-1-induced growth inhibition, apoptosis, ADCC and CDC in Raji cells, while induction of autophagy could accelerate cytotoxic effects of chLym-1 on Raji cells. Furthermore, chLym-1-induced autophagy can mediate apoptosis through Caspase 9 activation, demonstrating the tumor-suppressing role of autophagy in antilymphoma effects of chLym-1. Moreover, chLym-1 can activate several upstream signaling pathways of autophagy including Akt/mTOR and extracellular signal-regulated kinase 1/2 (Erk1/2). These results elucidate the critical role of autophagy in cytotoxicity of chLym-1 antibody and suggest a potential therapeutic strategy of NHL therapy by monoclonal antibody chLym-1 in combination with autophagy inducer.
Collapse
Affiliation(s)
- Jiajun Fan
- Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China
| | - Xian Zeng
- Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China
| | - Yubin Li
- Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China
- Key Laboratory for Microbiological Engineering of Agricultural Environment, Ministry of Agriculture, College of Life Science, Nanjing Agricultural University, Nanjing, Jiangsu, China
| | - Shaofei Wang
- Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China
| | - Ziyu Wang
- Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China
| | - Yun Sun
- Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China
| | - Hongjian Gao
- School of Medicine, Fudan University, Shanghai, China
| | - Guoping Zhang
- Institute of Biomedical Science, Fudan University, Shanghai, China
| | - Meiqing Feng
- Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China
- * E-mail: (DJ); (MF)
| | - Dianwen Ju
- Department of Biosynthesis, School of Pharmacy, Fudan University, Shanghai, China
- * E-mail: (DJ); (MF)
| |
Collapse
|
29
|
Zang L, He H, Xu Q, Yu Y, Zheng N, Liu W, Hayashi T, Tashiro SI, Onodera S, Ikejima T. Reactive oxygen species H2O2 and •OH, but not O2•(-) promote oridonin-induced phagocytosis of apoptotic cells by human histocytic lymphoma U937 cells. Int Immunopharmacol 2013; 15:414-23. [PMID: 23352441 DOI: 10.1016/j.intimp.2013.01.004] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2012] [Revised: 01/04/2013] [Accepted: 01/07/2013] [Indexed: 01/28/2023]
Abstract
We reported previously that phagocytosis of apoptotic cells by U937 cells was enhanced by the treatment with oridonin that showed high activity to induce the generation of reactive oxygen species (ROS) in many cells. ROS, important signaling molecules, are involved in the immune defenses, cell repair and proliferation. In this study, oridonin caused modest amount of ROS generation in U937 cells, with hydrogen peroxide (H2O2) and hydroxyl free radical (OH) as the major types. Meanwhile, H2O2 and OH were positive regulators involved in oridonin-enhanced engulfment of apoptotic cells through down-regulating mitochondrial membrane potential (MMP) and inducing autophagy. The ROS-mediated phagocytosis was independent of cellular adenosine triphosphate (ATP) levels. H2O2 and OH generation also activated phosphatidylinositol 3-kinases-Akt (PI3K-Akt) and phospholipase C γ-protein kinase C(PLC γ)-Ras-Raf-ERK signaling pathways, which were essential for oridonin-induced engulfment of apoptotic cells. Phagocytosis, the loss of MMP, autophagy and the activated signaling pathways were all suppressed by ROS scavenger N-acetyl-l-cysteine (NAC), H2O2 scavenger catalase or OH scavenger glutathione (GSH). However, superoxide anion (O2-) and its scavenger superoxide dismutase (SOD) did not significantly affect these oridonin-induced biological processes.
Collapse
Affiliation(s)
- Linghe Zang
- China-Japan Research Institute of Medical Pharmaceutical Sciences, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, PR China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
|
31
|
Liu Z, Ouyang L, Peng H, Zhang WZ. Oridonin: targeting programmed cell death pathways as an anti-tumour agent. Cell Prolif 2013; 45:499-507. [PMID: 23106297 DOI: 10.1111/j.1365-2184.2012.00849.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Oridonin, an active diterpenoid isolated from traditional Chinese herbal medicine, has drawn rising attention for its remarkable apoptosis- and autophagy-inducing activity and relevant molecular mechanisms in cancer therapy. Apoptosis is a well known type of cell death, whereas autophagy can play either pro-survival or pro-death roles in cancer cells. Accumulating evidence has recently revealed relationships between apoptosis and autophagy induced by oridonin; however, molecular mechanisms behind them remain to be discovered. In this review, we focus on highlighting updated research on oridonin-induced cell death signalling pathways implicated in apoptosis and autophagy, in many types of cancer. In addition, we further discuss cross-talk between apoptosis and autophagy induced by oridonin, in cancer. Taken together, these findings open new perspectives for further exploring oridonin as a potential anti-tumour agent targeting apoptosis and autophagy, in future anti-cancer therapeutics.
Collapse
Affiliation(s)
- Z Liu
- Department of Hepato-biliary Surgery, General Hospital of PLA, Beijing, China
| | | | | | | |
Collapse
|
32
|
Sun KW, Ma YY, Guan TP, Xia YJ, Shao CM, Chen LG, Ren YJ, Yao HB, Yang Q, He XJ. Oridonin induces apoptosis in gastric cancer through Apaf-1, cytochrome c and caspase-3 signaling pathway. World J Gastroenterol 2012; 18:7166-74. [PMID: 23326121 PMCID: PMC3544018 DOI: 10.3748/wjg.v18.i48.7166] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 09/21/2012] [Accepted: 10/16/2012] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect and mechanism of oridonin on the gastric cancer cell line HGC-27 in vitro.
METHODS: The inhibitory effect of oridonin on HGC-27 cells was detected using the 3-(4, 5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay. After treatment with 10 μg/mL oridonin for 24 h and 48 h, the cells were stained with acridine orange/ethidium bromide. The morphologic changes were observed under an inverted fluorescence microscope. DNA fragmentation (a hallmark of apoptosis) and lactate dehydrogenase activity were examined using DNA ladder assay and lactate dehydrogenase-release assay. After treated with oridonin (0, 1.25, 2.5, 5 and 10 μg/mL), HGC-27 cells were collected for anexin V-phycoerythrin and 7-amino-actinomycin D double staining and tested by flow cytometric analysis, and oridonin- induced apoptosis in HGC-27 cells was detected. After treatment with oridonin for 24 h, the effects of oridonin on expression of Apaf-1, Bcl-2, Bax, caspase-3 and cytochrome c were also analyzed using reverse-transcript polymerase chain reaction (RT-PCR) and Western blotting.
RESULTS: Oridonin significantly inhibited the proliferation of HGC-27 cells in a dose- and time-dependent manner. The inhibition rates of HGC-27 treated with four different concentrations of oridonin for 24 h (1.25, 2.5, 5 and 10 μg/mL) were 1.78% ± 0.36%, 4.96% ± 1.59%, 10.35% ± 2.76% and 41.6% ± 4.29%, respectively, which showed a significant difference (P < 0.05). The inhibition rates of HGC-27 treated with oridonin at the four concentrations for 48 h were 14.77% ± 4.21%, 21.57% ± 3.75%, 30.31% ± 4.91% and 61.19% ± 5.81%, with a significant difference (P < 0.05). The inhibition rates of HGC-27 treated with oridonin for 72 h at the four concentrations were 25.77% ± 4.85%, 31.86% ± 3.86%, 48.30% ± 4.16% and 81.80% ± 6.72%, with a significant difference (P < 0.05). Cells treated with oridonin showed typical apoptotic features with acridine orange/ethidium bromide staining. After treatment with oridonin, the cells became round, shrank, and developed small buds around the nuclear membrane while forming apoptotic bodies. Lactate dehydrogenase (LDH) release assay showed that after treated with 1.25 μg/mL and 20 μg/mL oridonin for 24 h, LDH release of HGC-27 caused by apoptosis increased from 22.94% ± 3.8% to 52.68% ± 2.4% (P < 0.001). However, the change in the release of LDH caused by necrosis was insignificant, suggesting that the major cause of oridonin-induced HGC-27 cell death was apoptosis. Flow cytometric analysis also revealed that oridonin induced significant apoptosis compared with the controls (P < 0.05). And the apoptosis rates of HGC-27 induced by the four different concentrations of oridonin were 5.3% ± 1.02%, 12.8% ± 2.53%, 28.5% ± 4.23% and 49.6% ± 3.76%, which were in a dose-dependent manner (P < 0.05). After treatment for 24 h, DNA ladder showed that oridonin induced a significant increase in DNA fragmentation in a dose-dependent manner. RT-PCR revealed that mRNA expression levels were up-regulated compared with the controls in caspase-3 (0.917 ± 0.103 vs 0.357 ± 0.019, P < 0.05), cytochrome c (1.429 ± 0.111 vs 1.002 ± 0.014, P < 0.05), Apaf-1 (0.688 ± 0.101 vs 0.242 ± 0.037, P < 0.05) and Bax (0.856 ± 0.101 vs 0.278 ± 0.027, P < 0.05) (P < 0.05), whereas down-regulated in Bcl-2 (0.085 ± 0.012 vs 0.175 ± 0.030, P < 0.05). Western blotting analysis also confirmed this result.
CONCLUSION: Apoptosis of HGC-27 induced by oridonin may be associated with differential expression of Apaf-1, caspase-3 and cytochrome c, which are highly dependent upon the mitochondrial pathway.
Collapse
|
33
|
Autophagy appears during the development of the mouse lower first molar. Histochem Cell Biol 2012; 139:109-18. [DOI: 10.1007/s00418-012-1016-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/03/2012] [Indexed: 12/19/2022]
|
34
|
Williams LR, Taylor GS. Autophagy and immunity - insights from human herpesviruses. Front Immunol 2012; 3:170. [PMID: 22783253 PMCID: PMC3389338 DOI: 10.3389/fimmu.2012.00170] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Accepted: 06/05/2012] [Indexed: 11/13/2022] Open
Abstract
The herpesviruses are a family of double-stranded DNA viruses that infect a wide variety of organisms. Having co-evolved with their hosts over millennia, herpesviruses have developed a large repertoire of mechanisms to manipulate normal cellular processes for their own benefit. Consequently, studies on these viruses have made important contributions to our understanding of fundamental biological processes. Here we describe recent research on the human herpesviruses that has contributed to our understanding of, and interactions between, viruses, autophagy, and the immune system. The ability of autophagy to degrade proteins located within the nucleus, the site of herpesvirus latency and replication, is also considered.
Collapse
Affiliation(s)
- Luke R Williams
- School of Cancer Sciences, University of Birmingham, Vincent Drive, Birmingham, UK
| | | |
Collapse
|
35
|
Zang L, He H, Ye Y, Liu W, Fan S, Tashiro SI, Onodera S, Ikejima T. Nitric oxide augments oridonin-induced efferocytosis by human histocytic lymphoma U937 cells via autophagy and the NF-κB-COX-2-IL-1β pathway. Free Radic Res 2012; 46:1207-19. [PMID: 22670565 DOI: 10.3109/10715762.2012.700515] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
We previously demonstrated that oridonin-induced autophagy enhanced efferocytosis (phagocytosis of apoptotic cells) by macrophage-like U937 cells through activation of the inflammatory pathways. In this study, exposure of U937 cells to 2.5 μM oridonin caused up-regulation of inducible nitric oxide synthase (iNOS) expression and continuous endogenous generation of nitric oxide (NO), which was reversed by pre-treatment with the inhibitors of nitric oxide synthase 1400 W (dihydrochloride) or L-NAME (hydrochloride). NO donor sodium nitroprusside (SNP) and efferocytosis irritant lipopolysaccharide (LPS) could also exert NO generation and iNOS expression. Moreover, oridonin-induced stimulation of efferocytosis was significantly suppressed by 1400 W or L-NAME. In addition, 1400 W or L-NAME impaired oridonin-induced autophagy. Inhibition of autophagy with 3-methyladenine (3MA) or Beclin-1 siRNA attenuated the uptake of apoptotic cells with a slight increase in the production of NO. The pro-inflammatory cytokine interleukin-1β (IL-1β) has been reported to be involved in oridonin-induced efferocytosis in U937 cells and interact with NO to contribute to inflammatory responses. 1400 W or L-NAME blocked the secretion of IL-1β and the activation of NF-κB and COX-2. Provision of SNP or LPS in place of oridonin resulted in the similar enhancement of efferocytosis, autophagy, the release of IL-1β and the expression of signal protein. NO augmented the oridonin-induced efferocytosis by mediating autophagy and activating the NF-κB-COX-2-IL-1β pathway. Inhibition of NF-κB or COX-2 in turn decreased the production of NO and the expression of iNOS. There exists a positive feedback loop between NO generation and NF-κB-COX-2-IL-1β pathway.
Collapse
Affiliation(s)
- Linghe Zang
- China-Japan Research Institute of Medical and Pharmaceutical Sciences, Shenyang Pharmaceutical University , 103 Wenhua Road, Shenyang , People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|